Resource

Submit Your Request Now

Submit Your Request Now

×

Top Advantages of Using DARTS for Drug Target Identification

The drug discovery process is a multi-stage journey that demands precision at every step, beginning with the accurate identification of molecular targets. Target-based drug discovery strategies rely heavily on the ability to pinpoint and validate the proteins that drive disease pathology. Failure to properly identify a drug's true biological target can lead to costly late-stage failures, off-target toxicities, and poor therapeutic efficacy.

Traditional target identification methods—such as affinity purification, yeast two-hybrid screening, and label-dependent pull-down assays—often suffer from technical limitations. These include the need for chemical modifications to the ligand, potential disruption of native protein structure, and the inability to capture transient or weak interactions. Moreover, these techniques may not perform well in complex biological systems, where proteins exist in dynamic, multi-component networks.

Drug Affinity Responsive Target Stability (DARTS) offers a paradigm shift by providing a label-free, non-biased method to detect ligand-protein interactions. Based on the principle that ligand binding stabilizes proteins against proteolytic degradation, DARTS enables researchers to identify direct molecular targets within their native environment without chemical alteration or immobilization of the compound. The technique is increasingly recognized as a vital component of modern drug discovery pipelines, particularly as the complexity of therapeutic landscapes grows.

In this article, we will explore the top advantages of using DARTS for drug target identification, highlighting why it is a preferred method for researchers seeking specificity, physiological relevance, and operational efficiency.

 Alt: Illustration of DARTS for drug target identification. (A) DARTS process overview. (B) FKBP12 incubation with drugs, followed by proteolysis. (C) mTOR inhibitor (E4) showing protected TOR fragments (purple arrow) and nonspecific band (*).The DARTS method for drug target identification. (A) DARTS scheme. (B) Proof of principle with FKBP12 and drugs. (C) DARTS with mTOR inhibitor (E4), showing protected TOR fragments (purple arrow) and nonspecific band (*) (Lomenick et al., 2009)

Unbiased Target Discovery Without the Need for Chemical Modification

One of the major limitations of traditional target identification techniques, such as affinity chromatography or photoaffinity labeling, is the requirement to chemically modify the ligand. These modifications—such as biotinylation, fluorescent tagging, or crosslinker attachment—are intended to facilitate detection or capture of the ligand-protein complex. However, chemical alterations can significantly distort the molecule's pharmacological properties, affecting binding affinity, specificity, and cellular permeability. This leads to a risk of missing authentic interactions or identifying artifacts that are irrelevant to the compound's true biological activity.

DARTS eliminates the need for any chemical modification of the ligand. Instead, it leverages the natural stabilization that occurs when a ligand binds to its target protein. Upon protease treatment, proteins that are bound to the ligand exhibit increased resistance to enzymatic degradation compared to unbound proteins. These protected proteins are then enriched and identified through proteomic analysis. This mechanism enables truly unbiased target discovery, as it captures all proteins affected by the ligand under physiologically relevant conditions.

The ability to maintain the ligand in its native, bioactive form is particularly valuable for the study of small molecules with delicate structures, such as natural products or covalent inhibitors, where even minor modifications could disrupt critical binding interactions. Moreover, because DARTS does not rely on affinity tags or capture steps, it is capable of detecting both high-affinity and moderate- to low-affinity interactions, expanding the spectrum of targets that can be uncovered.

An illustrative example comes from the discovery of the molecular targets of the natural product triptolide, where DARTS enabled the identification of its direct protein partners without the need for chemical conjugation. Such unbiased approaches are critical not only for primary target discovery but also for revealing secondary targets that may contribute to a compound's efficacy or toxicity profile.

Ultimately, DARTS empowers researchers with the ability to interrogate the full landscape of ligand-protein interactions in a manner that preserves biological relevance, facilitating a more comprehensive understanding of drug mechanisms.

Broad Applicability Across Different Molecule Types

Protein function is intimately tied to its three-dimensional structure. Subtle changes in folding, post-translational modifications, or interactions with other biomolecules can profoundly impact a protein's binding characteristics. In many conventional target identification methods—especially those involving protein purification, immobilization, or harsh lysis conditions—there is a substantial risk of altering the protein's native conformation. These alterations can obscure critical binding sites, disrupt multi-protein complexes, or even artificially create non-physiological interactions, leading to inaccurate identification of drug targets.

DARTS fundamentally addresses this problem by operating under non-denaturing conditions. The technique is typically conducted in native cell lysates or tissue homogenates, using mild buffers that preserve the tertiary and quaternary structures of proteins. This allows ligand binding events to occur within an environment that closely mirrors the natural intracellular milieu, maintaining essential factors like co-factors, chaperones, lipid interactions, and multi-subunit assemblies.

This preservation of physiological conformation offers several critical advantages:

  • Detection of conformationally sensitive interactions: Some ligand-binding sites are only exposed or functional when a protein adopts a specific folded state or assembles into a complex. DARTS captures these nuances.
  • Retention of dynamic protein complexes: Many cellular processes involve transient, regulated protein-protein interactions. DARTS can identify drug impacts on entire complexes rather than isolated proteins.
  • Post-translational modifications are maintained: Phosphorylation, ubiquitination, acetylation, and other modifications that influence protein activity or ligand binding are preserved during the assay.

For example, the heat shock protein HSP90 exhibits different conformations based on ATP binding status, which critically affects drug interactions. Using DARTS, researchers can study HSP90 inhibitors in a context that preserves these conformational states, leading to more accurate target validation compared to purified protein assays.

Moreover, because DARTS does not require overexpression systems, it avoids artifacts caused by non-physiological protein concentrations that could mask true affinity differences. The method enables target identification directly in disease-relevant models—such as patient-derived xenografts or organoids—thereby increasing the translational potential of the findings.

In short, by safeguarding the natural conformation and context of proteins, DARTS ensures that identified interactions are biologically meaningful and therapeutically actionable.

Compatibility with Downstream Proteomics Analysis

DARTS lies in its seamless integration with modern proteomics technologies, allowing researchers not only to detect stabilized proteins but also to comprehensively identify and quantify them on a global scale. This compatibility turns DARTS from a qualitative observation into a highly quantitative, systems-level tool for drug target discovery.

After DARTS treatment—where proteins are differentially protected from proteolysis in the presence of the ligand—samples can be subjected to gel-based separation for targeted analyses or, more powerfully, processed for mass spectrometry (MS)-based proteomics. In a standard workflow:

  • Protease-resistant proteins are digested into peptides.
  • These peptides are separated by liquid chromatography (LC).
  • Mass spectrometry (MS/MS) is used to identify and quantify them.
  • Bioinformatic pipelines interpret the data to pinpoint enriched targets.

Label-Free Quantification (LFQ) approaches allow relative comparison of protein abundance between treated and untreated samples without the need for additional chemical labeling. This is ideal for initial exploratory DARTS studies where the goal is broad target profiling.

For higher multiplexing and deeper quantitative accuracy, isobaric labeling methods such as Tandem Mass Tagging (TMT) or iTRAQ can be employed. These strategies enable simultaneous comparison of multiple experimental conditions (e.g., different drug concentrations, time points, or competitive binding assays) within a single mass spectrometry run, increasing throughput and minimizing technical variability.

Integration with proteomics offers several major benefits:

  • Unbiased target discovery: Thousands of proteins can be screened at once, enabling the identification of both expected and novel targets.
  • Quantitative dose-response profiling: Changes in stabilization levels at different drug concentrations can validate direct binding events.
  • Discovery of protein complexes: Proteins indirectly stabilized due to drug-induced complex formation can be detected and mapped.
  • Post-translational modification analysis: MS workflows allow simultaneous monitoring of changes in phosphorylation, ubiquitination, acetylation, and other modifications that may influence target engagement.

Recent innovations such as data-independent acquisition (DIA) proteomics further enhance DARTS by improving reproducibility, sensitivity, and coverage of low-abundance proteins, expanding the dynamic range of detectable targets.

For example, researchers using DARTS coupled with TMT-LC-MS/MS were able to uncover multiple off-target interactions of kinase inhibitors that were previously missed by traditional affinity methods, leading to better predictions of drug efficacy and toxicity profiles.

Thus, the synergy between DARTS and modern proteomics provides an unprecedented ability to map the complex interaction landscapes of small molecules within biological systems, offering a powerful platform for mechanism-of-action studies, drug repurposing projects, and biomarker discovery initiatives.

Minimal Material Requirement and Cost-Effectiveness

Resource efficiency is a critical consideration in drug discovery and academic research environments alike. Some target identification techniques, such as affinity chromatography or crosslinking-based methods, require large quantities of purified ligand, labeled reagents, or engineered cell lines, making them prohibitively expensive and impractical, especially during early discovery phases. DARTS offers a significant advantage by requiring minimal amounts of both ligand and protein material without sacrificing sensitivity or robustness.

Typically, DARTS assays can be performed using:

  • Microgram to low milligram quantities of total protein lysate
  • Micromolar to sub-micromolar concentrations of ligand
  • Common laboratory reagents (e.g., pronase or thermolysin for proteolysis)
  • Standard molecular biology equipment (e.g., centrifuges, electrophoresis systems)

The low material requirement makes DARTS particularly well-suited for situations where sample availability is limited, such as:

  • Screening rare or difficult-to-synthesize compounds
  • Investigating precious clinical samples (e.g., patient biopsies)
  • Profiling early-stage hits from phenotypic screens before full synthesis scale-up

In addition to low sample demands, the overall cost of a DARTS experiment is dramatically lower compared to techniques that rely on specialized hardware (e.g., surface plasmon resonance (SPR) instruments) or expensive consumables (e.g., biotinylated ligands, isotopic labels). The enzymes used for proteolysis in DARTS are inexpensive and broadly available, and the mass spectrometry analysis can often be integrated into existing core facility pipelines, further reducing operational costs.

Furthermore, because DARTS is based on simple comparative workflows (treated vs. untreated), experimental design and troubleshooting are relatively straightforward, minimizing personnel training costs and improving reproducibility across different laboratories.

An important downstream benefit of DARTS' efficiency is the ability to conduct multiple experiments in parallel—testing different ligands, concentrations, or biological models—without significantly increasing resource demands. This scalability allows for comprehensive profiling of compound libraries or optimization of lead candidates without disproportionate investment.

In resource-constrained settings, such as academic labs, small biotech companies, or preliminary drug discovery projects, DARTS provides an economically viable, high-value approach to early-stage target validation and deconvolution.

High Specificity and Sensitivity for Ligand-Target Interactions

Accurately identifying the true biological targets of small molecules requires methods that are both highly specific—distinguishing genuine interactions from background noise—and sensitive enough to detect low-abundance or transient binding events. DARTS excels on both fronts, offering a robust solution for uncovering ligand-protein interactions with a high degree of fidelity.

The core strength of DARTS in achieving specificity lies in its functional principle: only proteins that undergo a conformational change upon ligand binding exhibit increased resistance to proteolysis. This biochemical selectivity dramatically reduces false positives, as non-specifically associated or unbound proteins remain susceptible to enzymatic digestion and are therefore depleted from the final analysis. As a result, DARTS effectively filters out background interactions that often complicate interpretation in affinity-based or crosslinking-based approaches.

DARTS also demonstrates remarkable sensitivity, capable of detecting:

  • Low-affinity interactions that may be physiologically relevant but difficult to capture using traditional pull-down methods.
  • Allosteric binding events where ligand interaction induces stabilization at distant sites within a protein's structure.
  • Minor subpopulations of modified or conformationally distinct proteins, which could represent critical regulatory states within the cell.

Several refinements have further enhanced DARTS sensitivity and specificity:

  • Optimization of protease concentration ensures sufficient partial digestion to differentiate stabilized proteins without complete degradation.
  • Time-course studies allow researchers to fine-tune digestion windows to maximize detection of ligand-protected species.
  • Competitive binding experiments using excess unlabeled ligand can validate specific target engagement and rule out non-specific stabilization effects.

In application, DARTS has enabled researchers to uncover previously unknown secondary targets responsible for side effects or synergistic activities of therapeutics—findings that were not accessible through more conventional methods. For example, DARTS-based analysis revealed off-target kinase interactions of certain anti-cancer agents that helped explain unexpected clinical toxicities.

Importantly, by maintaining the endogenous protein-protein interaction network during the assay, DARTS can capture not only direct binders but also proximal interactors stabilized by drug engagement, providing a more comprehensive view of a compound's mechanism of action.

Facilitates Discovery of Off-Target Effects

Identifying off-target interactions is a crucial aspect of drug development, particularly for minimizing adverse side effects and improving therapeutic selectivity. While traditional biochemical assays often focus solely on validating a primary target, many clinical failures or post-approval withdrawals are later attributed to undetected off-target activities. DARTS provides an effective strategy for systematically uncovering these unintended interactions early in the development process.

Because DARTS does not rely on preselected targets or labeled ligands, it operates in an unbiased, proteome-wide manner, allowing researchers to simultaneously monitor stabilization across thousands of proteins. Ligand-induced stabilization is not restricted to the intended target; any protein that binds the drug and experiences increased resistance to proteolysis will be detected. This capacity makes DARTS an ideal platform for off-target profiling without prior assumptions.

Several important features enable DARTS to reveal off-target effects:

  • Proteome-wide sensitivity: High-coverage mass spectrometry combined with DARTS can detect low-abundance off-targets that might otherwise be missed.
  • Quantitative comparison: Degree of stabilization across targets can be compared, providing clues about binding affinity and functional relevance.
  • Competition assays: Excess unlabeled ligand or structurally related inactive compounds can help validate specific vs. non-specific binding events.

Real-world applications illustrate the power of DARTS for off-target discovery. For instance, studies with kinase inhibitors have revealed unexpected binding to non-kinase proteins, helping to explain complex pharmacological profiles. Similarly, DARTS has been used to uncover off-target interactions responsible for toxicity in cardiovascular and neurological systems, providing critical data for lead optimization or risk assessment.

By enabling early identification of off-target liabilities, DARTS supports de-risking strategies during preclinical development, informing structure-activity relationship (SAR) campaigns and enhancing the safety profile of therapeutic candidates. In the competitive landscape of drug discovery, where differentiation often hinges on subtle improvements in specificity and tolerability, integrating DARTS into the target validation workflow can offer a decisive advantage.

Enables Studies in Complex Biological Mixtures

One of the persistent challenges in drug target identification is maintaining biological relevance when transitioning from simplified in vitro systems to the complex environments of real biological tissues and cells. Many traditional techniques, such as immobilized target assays or recombinant overexpression systems, sacrifice physiological complexity for experimental convenience—potentially missing critical context-dependent interactions.

DARTS, by contrast, excels in preserving biological complexity, allowing researchers to study ligand-target interactions directly within heterogeneous, highly intricate biological samples.

Because DARTS operates without the need for chemical modification, immobilization, or excessive purification, it can be applied to native cell lysates, tissue extracts, or even primary clinical samples with minimal perturbation of the proteome. This capacity brings several distinct advantages:

  • Maintaining endogenous protein levels: Studying proteins at their native expression levels avoids artifacts caused by overexpression or purification-induced misfolding.
  • Capturing complex protein-protein interaction networks: In tissues or whole-cell lysates, many proteins exist as part of dynamic multi-protein complexes. DARTS can reveal how small molecules interact with these complexes, not just isolated proteins.
  • Preserving post-translational modifications: Many biologically relevant binding events depend on the phosphorylation, acetylation, glycosylation, or ubiquitination states of target proteins. DARTS preserves these modifications, enabling detection of context-specific interactions.
  • Application to disease-relevant samples: Importantly, DARTS can be performed on patient-derived samples, such as tumor biopsies or diseased tissues, enabling identification of disease-specific drug targets or biomarkers under clinically relevant conditions.

For instance, DARTS has been successfully applied to brain tissue lysates to identify novel neuroprotective targets of small molecules in models of neurodegenerative disease. In oncology research, DARTS has been used on primary tumor specimens to reveal differential target engagement patterns compared to normal tissues, informing personalized therapy strategies.

Furthermore, because DARTS tolerates the complexity of biological mixtures, it opens possibilities for studying ligand interactions within whole organoids, complex co-cultures, or microenvironment-mimicking systems—models that better replicate human physiology than traditional monocultures.

Reference

  1. Lomenick, Brett, et al. "Target identification using drug affinity responsive target stability (DARTS)." Proceedings of the National Academy of Sciences 106.51 (2009): 21984-21989. https://doi.org/10.1073/pnas.0910040106
* For Research Use Only. Not for use in diagnostic procedures.
Our customer service representatives are available 24 hours a day, 7 days a week. Inquiry

From Our Clients

Online Inquiry

Please submit a detailed description of your project. We will provide you with a customized project plan to meet your research requests. You can also send emails directly to for inquiries.

* Email
Phone
* Service & Products of Interest
Services Required and Project Description
* Verification Code
Verification Code

Great Minds Choose Creative Proteomics